Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Development ; 145(9)2018 04 30.
Article in English | MEDLINE | ID: mdl-29712669

ABSTRACT

The architecture of individual cells and cell collectives enables functional specification, a prominent example being the formation of epithelial tubes that transport fluid or gas in many organs. The intrahepatic bile ducts (IHBDs) form a tubular network within the liver parenchyma that transports bile to the intestine. Aberrant biliary 'neoductulogenesis' is also a feature of several liver pathologies including tumorigenesis. However, the mechanism of biliary tube morphogenesis in development or disease is not known. Elimination of the neurofibromatosis type 2 protein (NF2; also known as merlin or neurofibromin 2) causes hepatomegaly due to massive biliary neoductulogenesis in the mouse liver. We show that this phenotype reflects unlimited biliary morphogenesis rather than proliferative expansion. Our studies suggest that NF2 normally limits biliary morphogenesis by coordinating lumen expansion and cell architecture. This work provides fundamental insight into how biliary fate and tubulogenesis are coordinated during development and will guide analyses of disease-associated and experimentally induced biliary pathologies.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Cell Proliferation/physiology , Neurofibromin 2/metabolism , Organogenesis/physiology , Animals , Bile Ducts, Intrahepatic/pathology , Gene Deletion , Hepatomegaly/embryology , Hepatomegaly/genetics , Hepatomegaly/pathology , Mice , Mice, Knockout , Neurofibromin 2/genetics
2.
Genes Dev ; 30(17): 1971-90, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27664237

ABSTRACT

IKAROS is required for the differentiation of highly proliferative pre-B-cell precursors, and loss of IKAROS function indicates poor prognosis in precursor B-cell acute lymphoblastic leukemia (B-ALL). Here we show that IKAROS regulates this developmental stage by positive and negative regulation of superenhancers with distinct lineage affiliations. IKAROS defines superenhancers at pre-B-cell differentiation genes together with B-cell master regulators such as PAX5, EBF1, and IRF4 but is required for a highly permissive chromatin environment, a function that cannot be compensated for by the other transcription factors. IKAROS is also highly enriched at inactive enhancers of genes normally expressed in stem-epithelial cells. Upon IKAROS loss, expression of pre-B-cell differentiation genes is attenuated, while a group of extralineage transcription factors that are directly repressed by IKAROS and depend on EBF1 relocalization at their enhancers for expression is induced. LHX2, LMO2, and TEAD-YAP1, normally kept separate from native B-cell transcription regulators by IKAROS, now cooperate directly with them in a de novo superenhancer network with its own feed-forward transcriptional reinforcement. Induction of de novo superenhancers antagonizes Polycomb repression and superimposes aberrant stem-epithelial cell properties in a B-cell precursor. This dual mechanism of IKAROS regulation promotes differentiation while safeguarding against a hybrid stem-epithelial-B-cell phenotype that underlies high-risk B-ALL.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic/physiology , Epithelial Cells/cytology , Gene Expression Regulation, Leukemic , Ikaros Transcription Factor/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Precursor Cells, B-Lymphoid/cytology , Animals , Epigenesis, Genetic , Epithelial Cells/pathology , Ikaros Transcription Factor/genetics , Mice , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Cell ; 165(6): 1401-1415, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27180906

ABSTRACT

Chromatin remodeling proteins are frequently dysregulated in human cancer, yet little is known about how they control tumorigenesis. Here, we uncover an epigenetic program mediated by the NAD(+)-dependent histone deacetylase Sirtuin 6 (SIRT6) that is critical for suppression of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal malignancies. SIRT6 inactivation accelerates PDAC progression and metastasis via upregulation of Lin28b, a negative regulator of the let-7 microRNA. SIRT6 loss results in histone hyperacetylation at the Lin28b promoter, Myc recruitment, and pronounced induction of Lin28b and downstream let-7 target genes, HMGA2, IGF2BP1, and IGF2BP3. This epigenetic program defines a distinct subset with a poor prognosis, representing 30%-40% of human PDAC, characterized by reduced SIRT6 expression and an exquisite dependence on Lin28b for tumor growth. Thus, we identify SIRT6 as an important PDAC tumor suppressor and uncover the Lin28b pathway as a potential therapeutic target in a molecularly defined PDAC subset. PAPERCLIP.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , RNA-Binding Proteins/genetics , Sirtuins/genetics , Acetylation , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Female , Genes, ras , Histones/metabolism , Humans , Male , Mice , Mice, Knockout , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism
5.
J Natl Cancer Inst ; 107(10)2015 Oct.
Article in English | MEDLINE | ID: mdl-26293574

ABSTRACT

BACKGROUND: Huntingtin (HTT) is mutated in Huntington's disease but is ubiquitously expressed, and mutant HTT influences cancer progression. We investigated wild-type HTT function during breast cancer. METHODS: We analyzed HTT and ZO1 expression as well as the HTT phosphoserine 421-activated form (S421-P-HTT) in human breast cancer tissues by quantitative reverse transcription polymerase chain reaction and immunohistochemistry. We performed in vitro migration and invasion assays as well as in vivo tail vein injections of the metastatic 4T1 cells in BALB/c mice (n = 11 per group). We analyzed tumor progression in knock-in mice with modified S421 crossed with the MMTV-PyVT mammary cancer model (at least n = 12 per group). Data were analyzed with unpaired t tests, analysis of variance, Pearson or Spearman correlation, and Mann Whitney or Kruskal-Wallis tests. All statistical tests were two-sided. RESULTS: Levels of HTT and of S421-P-HTT are abnormally low in poorly differentiated and metastatic human breast cancers. HTT expression is downregulated in invasive compared with in situ carcinoma (P < .001). In BALB/c mice, silencing of HTT promotes lung colonization by a metastatic mammary cancer cell line (P = .005) and S421-unphosphorylatable-HTT accelerates cancer progression. HTT interacts with ZO1 and regulates both its expression and its localization to tight junctions. In human breast tumors, the patterns of HTT and ZO1 expression are similar (Pearson correlation coefficient = 0.66, P < .001). CONCLUSIONS: HTT may inhibit breast tumor dissemination through maintenance of ZO1 at tight junctions. Downregulation of HTT transcript and protein levels is a prognostic factor for poor prognosis and metastasis development.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Silencing , Nerve Tissue Proteins/metabolism , Zonula Occludens-1 Protein/metabolism , Animals , Breast Neoplasms/pathology , Cell Movement , Disease Progression , Down-Regulation , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Huntingtin Protein , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Phosphorylation , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured
6.
Nature ; 524(7565): 361-5, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26168401

ABSTRACT

Activation of cellular stress response pathways to maintain metabolic homeostasis is emerging as a critical growth and survival mechanism in many cancers. The pathogenesis of pancreatic ductal adenocarcinoma (PDA) requires high levels of autophagy, a conserved self-degradative process. However, the regulatory circuits that activate autophagy and reprogram PDA cell metabolism are unknown. Here we show that autophagy induction in PDA occurs as part of a broader transcriptional program that coordinates activation of lysosome biogenesis and function, and nutrient scavenging, mediated by the MiT/TFE family of transcription factors. In human PDA cells, the MiT/TFE proteins--MITF, TFE3 and TFEB--are decoupled from regulatory mechanisms that control their cytoplasmic retention. Increased nuclear import in turn drives the expression of a coherent network of genes that induce high levels of lysosomal catabolic function essential for PDA growth. Unbiased global metabolite profiling reveals that MiT/TFE-dependent autophagy-lysosome activation is specifically required to maintain intracellular amino acid pools. These results identify the MiT/TFE proteins as master regulators of metabolic reprogramming in pancreatic cancer and demonstrate that transcriptional activation of clearance pathways converging on the lysosome is a novel hallmark of aggressive malignancy.


Subject(s)
Autophagy/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Lysosomes/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Amino Acids/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Energy Metabolism , Female , Heterografts , Homeostasis , Humans , Lysosomes/genetics , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Transcription, Genetic
7.
Cell ; 161(7): 1553-65, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26073944

ABSTRACT

Hematopoietic stem cells (HSCs) reside in hypoxic niches within bone marrow and cord blood. Yet, essentially all HSC studies have been performed with cells isolated and processed in non-physiologic ambient air. By collecting and manipulating bone marrow and cord blood in native conditions of hypoxia, we demonstrate that brief exposure to ambient oxygen decreases recovery of long-term repopulating HSCs and increases progenitor cells, a phenomenon we term extraphysiologic oxygen shock/stress (EPHOSS). Thus, true numbers of HSCs in the bone marrow and cord blood are routinely underestimated. We linked ROS production and induction of the mitochondrial permeability transition pore (MPTP) via cyclophilin D and p53 as mechanisms of EPHOSS. The MPTP inhibitor cyclosporin A protects mouse bone marrow and human cord blood HSCs from EPHOSS during collection in air, resulting in increased recovery of transplantable HSCs. Mitigating EPHOSS during cell collection and processing by pharmacological means may be clinically advantageous for transplantation.


Subject(s)
Bone Marrow , Fetal Blood/cytology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Female , Hematopoietic Stem Cell Transplantation/instrumentation , Hematopoietic Stem Cells/cytology , Humans , Hypoxia , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Cell Rep ; 10(10): 1692-1707, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25772357

ABSTRACT

Defective Hippo/YAP signaling in the liver results in tissue overgrowth and development of hepatocellular carcinoma (HCC). Here, we uncover mechanisms of YAP-mediated hepatocyte reprogramming and HCC pathogenesis. YAP functions as a rheostat in maintaining metabolic specialization, differentiation, and quiescence within the hepatocyte compartment. Increased or decreased YAP activity reprograms subsets of hepatocytes to different fates associated with deregulation of the HNF4A, CTNNB1, and E2F transcriptional programs that control hepatocyte quiescence and differentiation. Importantly, treatment with small interfering RNA-lipid nanoparticles (siRNA-LNPs) targeting YAP restores hepatocyte differentiation and causes pronounced tumor regression in a genetically engineered mouse HCC model. Furthermore, YAP targets are enriched in an aggressive human HCC subtype characterized by a proliferative signature and absence of CTNNB1 mutations. Thus, our work reveals Hippo signaling as a key regulator of the positional identity of hepatocytes, supports targeting of YAP using siRNA-LNPs as a paradigm of differentiation-based therapy, and identifies an HCC subtype that is potentially responsive to this approach.

9.
Cell Rep ; 9(2): 495-503, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25373897

ABSTRACT

Hippo signaling is a tumor-suppressor pathway involved in organ size control and tumorigenesis through the inhibition of YAP and TAZ. Here, we show that energy stress induces YAP cytoplasmic retention and S127 phosphorylation and inhibits YAP transcriptional activity and YAP-dependent transformation. These effects require the central metabolic sensor AMP-activated protein kinase (AMPK) and the upstream Hippo pathway components Lats1/Lats2 and angiomotin-like 1 (AMOTL1). Furthermore, we show that AMPK directly phosphorylates S793 of AMOTL1. AMPK activation stabilizes and increases AMOTL1 steady-state protein levels, contributing to YAP inhibition. The phosphorylation-deficient S793Ala mutant of AMOTL1 showed a shorter half-life and conferred resistance to energy-stress-induced YAP inhibition. Our findings link energy sensing to the Hippo-YAP pathway and suggest that YAP may integrate spatial (contact inhibition), mechanical, and metabolic signals to control cellular proliferation and survival.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenylate Kinase/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stress, Physiological , Amino Acid Sequence , Angiomotins , Energy Metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , Membrane Proteins/genetics , Molecular Sequence Data , Mutation, Missense , Phosphorylation , Protein Stability , Transcription Factors , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
10.
Nature ; 513(7516): 110-4, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25043045

ABSTRACT

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are among the most common genetic alterations in intrahepatic cholangiocarcinoma (IHCC), a deadly liver cancer. Mutant IDH proteins in IHCC and other malignancies acquire an abnormal enzymatic activity allowing them to convert α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), which inhibits the activity of multiple αKG-dependent dioxygenases, and results in alterations in cell differentiation, survival, and extracellular matrix maturation. However, the molecular pathways by which IDH mutations lead to tumour formation remain unclear. Here we show that mutant IDH blocks liver progenitor cells from undergoing hepatocyte differentiation through the production of 2HG and suppression of HNF-4α, a master regulator of hepatocyte identity and quiescence. Correspondingly, genetically engineered mouse models expressing mutant IDH in the adult liver show an aberrant response to hepatic injury, characterized by HNF-4α silencing, impaired hepatocyte differentiation, and markedly elevated levels of cell proliferation. Moreover, IDH and Kras mutations, genetic alterations that co-exist in a subset of human IHCCs, cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic IHCC. These studies provide a functional link between IDH mutations, hepatic cell fate, and IHCC pathogenesis, and present a novel genetically engineered mouse model of IDH-driven malignancy.


Subject(s)
Bile Duct Neoplasms/pathology , Cell Differentiation/genetics , Cholangiocarcinoma/pathology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocytes/pathology , Isocitrate Dehydrogenase/genetics , Mutant Proteins/metabolism , Animals , Bile Duct Neoplasms/enzymology , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/enzymology , Bile Ducts, Intrahepatic/pathology , Cell Division/genetics , Cell Lineage/genetics , Cholangiocarcinoma/enzymology , Cholangiocarcinoma/genetics , Disease Models, Animal , Female , Glutarates/metabolism , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Male , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutation/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Stem Cells/pathology , ras Proteins/genetics , ras Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 111(30): E3091-100, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024225

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is the most lethal of common human malignancies, with no truly effective therapies for advanced disease. Preclinical studies have suggested a therapeutic benefit of targeting the Hedgehog (Hh) signaling pathway, which is activated throughout the course of PDA progression by expression of Hh ligands in the neoplastic epithelium and paracrine response in the stromal fibroblasts. Clinical trials to test this possibility, however, have yielded disappointing results. To further investigate the role of Hh signaling in the formation of PDA and its precursor lesion, pancreatic intraepithelial neoplasia (PanIN), we examined the effects of genetic or pharmacologic inhibition of Hh pathway activity in three distinct genetically engineered mouse models and found that Hh pathway inhibition accelerates rather than delays progression of oncogenic Kras-driven disease. Notably, pharmacologic inhibition of Hh pathway activity affected the balance between epithelial and stromal elements, suppressing stromal desmoplasia but also causing accelerated growth of the PanIN epithelium. In striking contrast, pathway activation using a small molecule agonist caused stromal hyperplasia and reduced epithelial proliferation. These results indicate that stromal response to Hh signaling is protective against PDA and that pharmacologic activation of pathway response can slow tumorigenesis. Our results provide evidence for a restraining role of stroma in PDA progression, suggesting an explanation for the failure of Hh inhibitors in clinical trials and pointing to the possibility of a novel type of therapeutic intervention.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Mice , Mice, Knockout , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
12.
J Clin Invest ; 124(1): 285-96, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24334454

ABSTRACT

Alveolar rhabdomyosarcoma (aRMS) is an aggressive sarcoma of skeletal muscle characterized by expression of the paired box 3-forkhead box protein O1 (PAX3-FOXO1) fusion oncogene. Despite its discovery nearly two decades ago, the mechanisms by which PAX3-FOXO1 drives tumor development are not well characterized. Previously, we reported that PAX3-FOXO1 supports aRMS initiation by enabling bypass of cellular senescence checkpoints. We have now found that this bypass occurs in part through PAX3-FOXO1-mediated upregulation of RASSF4, a Ras-association domain family (RASSF) member. RASSF4 expression was upregulated in PAX3-FOXO1-positive aRMS cell lines and tumors. Enhanced RASSF4 expression promoted cell cycle progression, senescence evasion, and tumorigenesis through inhibition of the Hippo pathway tumor suppressor MST1. We also found that the downstream Hippo pathway target Yes-associated protein 1 (YAP), which is ordinarily restrained by Hippo signaling, was upregulated in RMS tumors. These data suggest that Hippo pathway dysfunction promotes RMS. This work provides evidence for Hippo pathway suppression in aRMS and demonstrates a progrowth role for RASSF4. Additionally, we identify a mechanism used by PAX3-FOXO1 to inhibit MST1 signaling and promote tumorigenesis in aRMS.


Subject(s)
Carcinogenesis/metabolism , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Animals , Base Sequence , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Cellular Senescence , Drosophila , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Humans , Mice , Mice, SCID , Molecular Sequence Data , Myoblasts/physiology , Neoplasm Transplantation , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcriptome , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Nat Cell Biol ; 14(12): 1322-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143395

ABSTRACT

Organ development is a complex process governed by the interplay of several signalling pathways that have critical functions in the regulation of cell growth and proliferation. Over the past years, the Hippo pathway has emerged as a key regulator of organ size. Perturbation of this pathway has been shown to play important roles in tumorigenesis. YAP, the main downstream target of the mammalian Hippo pathway, promotes organ growth, yet the underlying molecular mechanism of this regulation remains unclear. Here we provide evidence that YAP activates the mammalian target of rapamycin (mTOR), a major regulator of cell growth. We have identified the tumour suppressor PTEN, an upstream negative regulator of mTOR, as a critical mediator of YAP in mTOR regulation. We demonstrate that YAP downregulates PTEN by inducing miR-29 to inhibit PTEN translation. Last, we show that PI(3)K­mTOR is a pathway modulated by YAP to regulate cell size, tissue growth and hyperplasia. Our studies reveal a functional link between Hippo and PI(3)K­mTOR, providing a molecular basis for the coordination of these two pathways in organ size regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepatocyte Growth Factor/metabolism , MicroRNAs/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line , Chromatin Immunoprecipitation , Chromones/pharmacology , Flow Cytometry , Hepatocyte Growth Factor/genetics , High-Throughput Nucleotide Sequencing , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Morpholines/pharmacology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Serine-Threonine Kinase 3 , YAP-Signaling Proteins
14.
Semin Cell Dev Biol ; 23(7): 770-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22898666

ABSTRACT

The "Hippo" signaling pathway has emerged as a major regulator of cell proliferation and survival in metazoans. The pathway, as delineated by genetic and biochemical studies in Drosophila, consists of a kinase cascade regulated by cell-cell contact and cell polarity that inhibits the transcriptional coactivator Yorkie and its proliferative, anti-differentiation, antiapoptotic transcriptional program. The core pathway components are the GC kinase Hippo, which phosphorylates the noncatalytic polypeptide Mats/Mob1 and, with the assistance of the scaffold protein Salvador, phosphorylates the ndr-family kinase Lats. In turn phospho-Lats, after binding to phospho-Mats, autoactivates and phosphorylates Yorkie, resulting in its nuclear exit. Hippo also uses the scaffold protein Furry and a different Mob protein to control another ndr-like kinase, the morphogenetic regulator Tricornered. Architecturally homologous kinase cascades consisting of a GC kinase, a Mob protein, a scaffolding polypeptide and an ndr-like kinase are well described in yeast; in Saccharomyces cerevisiae, e.g., the MEN pathway promotes mitotic exit whereas the RAM network, using a different GC kinase, Mob protein, scaffold and ndr-like kinase, regulates cell polarity and morphogenesis. In mammals, the Hippo orthologs Mst1 and Mst2 utilize the Salvador ortholog WW45/Sav1 and other scaffolds to regulate the kinases Lats1/Lats2 and ndr1/ndr2. As in Drosophila, murine Mst1/Mst2, in a redundant manner, negatively regulate the Yorkie ortholog YAP in the epithelial cells of the liver and gut; loss of both Mst1 and Mst2 results in hyperproliferation and tumorigenesis that can be largely negated by reduction or elimination of YAP. Despite this conservation, considerable diversification in pathway composition and regulation is already evident; in skin, e.g., YAP phosphorylation is independent of Mst1Mst2 and Lats1Lats2. Moreover, in lymphoid cells, Mst1/Mst2, under the control of the Rap1 GTPase and independent of YAP, promotes integrin clustering, actin remodeling and motility while restraining the proliferation of naïve T cells. This review will summarize current knowledge of the structure and regulation of the kinases Hippo/Mst1&2, their noncatalytic binding partners, Salvador and the Rassf polypeptides, and their major substrates Warts/Lats1&2, Trc/ndr1&2, Mats/Mob1 and FOXO.


Subject(s)
Protein Kinases/metabolism , Signal Transduction , Animals , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitosis , Substrate Specificity
15.
J Exp Med ; 206(4): 833-47, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19349462

ABSTRACT

Neuroblastoma (NB), the most frequent solid tumor of early childhood, is diagnosed as a disseminated disease in >60% of cases, and several lines of evidence support the resistance to apoptosis as a prerequisite for NB progression. We show that autocrine production of netrin-1, a multifunctional laminin-related molecule, conveys a selective advantage in tumor growth and dissemination in aggressive NB, as it blocks the proapoptotic activity of the UNC5H netrin-1 dependence receptors. We show that such netrin-1 up-regulation is a potential marker for poor prognosis in stage 4S and, more generally, in NB stage 4 diagnosed infants. Moreover, we propose that interference with the netrin-1 autocrine loop in malignant neuroblasts could represent an alternative therapeutic strategy, as disruption of this loop triggers in vitro NB cell death and inhibits NB metastasis in avian and mouse models.


Subject(s)
Nerve Growth Factors/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Tumor Suppressor Proteins/genetics , Cell Death , Cell Line, Tumor , Cell Survival , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , Infant , Infant, Newborn , Neoplasm Staging , Netrin-1 , Neuroblastoma/mortality , Prognosis , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survivors , Up-Regulation
16.
Expert Opin Ther Targets ; 12(8): 995-1007, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18620521

ABSTRACT

BACKGROUND: Netrin-1 belongs to a family of secreted proteins that act as migration and adhesion cues in the developing CNS and in a number of non-neural tissues. Netrin-1 is the ligand of deleted in colorectal cancer (DCC) and the uncoordinated family member 5 (UNC5) orthologues of the dependence receptor family. Over the past ten years, a novel mechanism has emerged, that a receptor unoccupied by its ligand is not necessarily inactive. Rather, such a receptor can mediate two signalling pathways, depending on whether it is bound to its ligand or not. In the absence of ligand, an active signalling pathway results in cell death through apoptosis. OBJECTIVE: Coupled netrin-1 receptors have been shown to regulate diverse processes such as maintenance, integrity, migration and renewal of many tissues. We propose that netrin-1 receptors can regulate tumour development. METHODS: We review the properties of netrin-1 and present netrin-1 receptors as regulators of tumourigenesis. RESULTS/CONCLUSION: Netrin-1 and its receptors are unexplored critical targets in cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Nerve Growth Factors/metabolism , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Humans , Netrin Receptors , Netrin-1
17.
Proc Natl Acad Sci U S A ; 105(12): 4850-5, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18353983

ABSTRACT

Netrin-1, an axon navigation cue was proposed to play a crucial role during colorectal tumorigenesis by regulating apoptosis. The netrin-1 receptors DCC and UNC5H were shown to belong to the family of dependence receptors that share the ability to induce apoptosis in the absence of their ligands. Such a trait confers on these receptors a tumor suppressor activity. Expression of one of these dependence receptors at the surface of a tumor cell is indeed speculated to render this cell dependent on ligand availability for its survival, hence inhibiting uncontrolled cell proliferation or metastasis. Consequently, it is a selective advantage for a tumor cell to lose this dependence receptor activity, as previously described with losses of DCC and UNC5H expression in human cancers. However, the model predicts that a similar advantage may be obtained by gaining autocrine expression of the ligand. We describe here that, unlike human nonmetastatic breast tumors, a large fraction of metastatic breast cancers overexpress netrin-1. Moreover, we show that netrin-1-expressing mammary metastatic tumor cell lines undergo apoptosis when netrin-1 expression is experimentally decreased or when decoy soluble receptor ectodomains are added. Such treatments prevent metastasis formation both in a syngenic mouse model of lung colonization of a mammary cancer cell line and in a model of spontaneous lung metastasis of xenografted human breast tumor. Thus, netrin-1 expression observed in a large fraction of human metastatic breast tumors confers a selective advantage for tumor cell survival and potentially represents a promising target for alternative anticancer therapeutic strategies.


Subject(s)
Breast Neoplasms/pathology , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Autocrine Communication , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Cell Death , Cell Line, Tumor , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Nerve Growth Factors/genetics , Netrin-1 , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...